Abstract
No common malignancy is as rapidly and inevitably fatal as pancreatic ductal adenocarcinoma (PDA). This grim fact has driven substantial research efforts into this disease in recent decades. Unfortunately, the investment has yet to result in a meaningful increase in 5-year survival. This has prompted many pancreatic cancer researchers and advocates to redouble their efforts, but also requires one to step back and ask why the previous efforts were lacking and to consider why pancreatic cancer is so difficult to treat. The difficulties are legion. PDA is characterized by an insidious clinical syndrome, but is rarely diagnosed at a time when surgical resection is feasible. We lack markers of early detection and screening programs remain unproven even in high risk populations. The location of the tumor in the retroperitoneum, the advanced age of patients, and the systemic effects of disease limit the options for local therapy. Chemotherapy may provide a small benefit, but most efforts to improve on the current regimens consistently and stubbornly fail in advanced clinical trials. The molecular and cellular features of ductal pancreatic tumors are aggressive and underlay multiple levels of therapeutic resistance. Non-cell-autonomous features including stromal proliferation, reduced vascular density and immune suppression also contribute to therapeutic resistance. Growing awareness of these the fundamental features of PDA has begun to guide ongoing research efforts. Clinical trials are now specifically targeting these tumor properties and actively focusing on the therapeutic implications of tumor stroma. As reviewed here, reflecting on the fundamental question of why pancreatic cancer is so difficult to treat is a necessary and informative exercise that will aid our efforts to improve patient outcomes. These efforts will lead to improvements in clinical trial design, expand our focus to include the molecular and histologic implications of novel treatment paradigms, and ultimately change the lives of our patients.
Keywords: pancreatic cancer, chemotherapy resistance, tumor desmoplasia
Introduction
In the modern era of cancer research, pancreatic ductal adenocarcinoma (PDA) has proven to be among the most unyielding of adversaries. The oncology community has expended its entire arsenal at this disease with little effect: the 5-year survival rate has ticked up to 6% over the past 40 years, but nearly all diagnosed patients ultimately succumb to the disease. An estimated 37,390 people will die of pancreatic cancer in the US in 2012 [Siegel et al. 2012] with a similar pattern in the rest of the developed world [Jemal et al. 2011]. Over 80% of them will be found to have unresectable tumors at diagnosis [Stathis and Moore, 2010], giving them an expected overall survival of just 6 months. There are few therapeutic options for these patients and the most efficacious are also the most burdensome. Those who do undergo surgery improve their overall survival compared with patients of a similar stage by about 10 months [Bilimoria et al. 2007], but must tolerate significant morbidity and face almost inevitable recurrence. Given the slow progress against this disease, one must ask the question ‘why is pancreatic cancer so hard to treat?’.
The particular problem of pancreatic cancer is multifactorial in its nature. The patient population in PDA is predominantly elderly and in poor overall health. There is no simple early detection method for pancreatic cancer and the earliest indications of disease are nonspecific. The tumor itself has its own peculiarities. For example, it has become apparent that PDA metastasizes microscopically early in the disease course, limiting the effectiveness of local therapies such as surgery and radiation. At the cellular level, the actual neoplastic epithelial cells at the heart of the disease harbor some of the most profoundly oncogenic alterations known to biology, and these are found at unusually high frequencies in PDA. In addition to driving growth and promoting cell survival, these alterations alter the metabolism of pancreatic cancer to one that can better support the manufacture of new cellular components. Layered on top of these high penetrance mutations is a host of rare alterations that are found in effectively unique combinations in each patient. The extent of genetic alterations in pancreatic tumors bears witness to a genomic instability phenotype that appears to play a significant role in the biology of PDA and implies an ability to rapidly develop acquired resistance to therapies that do manage to provoke an initial response. In addition to features of the tumor epithelium, PDA harbors a dense, desmoplastic stroma that can serve to limit the delivery of agents to tumors and foreshadows an incredibly complex interplay of intercellular signals that confound our ability to study the disease in vitro. Certain cell types within this stroma construct an immune-suppressed microenvironment that prevents the local immune system from clearing the tumor. Finally, PDA manifests as a syndrome, not just a mass, with systemic comorbidities that have a profoundly negative impact on quality of life.
Together, these raw observations paint a grim picture of the battle against pancreatic cancer that has at times led to a sense of nihilism. In reality, there are many signs that the research efforts of the past few decades have altered the momentum of this battle. Each of the challenges listed above has, in recent years, been the subject of intense research, leading to new ideas that are now being developed in the lab and in the clinic. For example, an understanding of the dynamics of drug delivery in PDA has led to a focus on targeted agents with desirable pharmacological properties. Another approach is to target the tumor stroma directly in order to facilitate the delivery of genotoxic agents or relieve local immune suppression. Other agents take advantage of the hypoxic microenvironment conferred by the desmoplastic stroma, or specific metabolic dependencies. Furthermore, decades of failed trials have led to improvements in clinical trial design and in the diagnostic and interventional techniques used in patients. By addressing the manifold difficulties that underpin the challenge of pancreatic cancer, a new sense of optimism is apparent. These barriers are surmountable and the nascent efforts to address them will ultimately be reflected in improved patient outcomes.
Patient population and diagnosis
Pancreatic ductal adenocarcinoma is largely a disease of old age, with an average age of diagnosis of 71 years. Yet the presenting symptoms are nonspecific such as weight loss and abdominal pain [Bakkevold et al. 1992]. This population of patients (and their general practitioners) is accustomed to aches and pains, and so in most cases, the earliest signs of malignancy go unnoticed; a high level of perception is required to avoid delays in diagnostic workup. Furthermore, in contrast to breast, prostate, melanoma and testicular cancers, there are no simple examinations that can elevate the level of suspicion: the pancreas is too deep to palpate and there is no specific blood test available for PDA. Other symptoms at diagnosis can include new onset of diabetes [Chari et al. 2005], unexplained jaundice [Porta et al. 2005] and unprovoked thrombosis [Khorana and Fine, 2004]; the most specific of these is unexplained painless jaundice, but many other explanations are possible. Thus, by the time that a patient seeks medical advice and their GP successfully navigates the diagnostic maze, often many months have passed and the patient’s condition has further deteriorated.
PDA is associated with a syndrome of comorbidities that affect patients’ overall health and in some cases can be life threatening. Symptoms related to pain [Porta et al. 2005] and depression [Kelsen et al. 1995] are components of this syndrome and are often present at the time of diagnosis, but become more severe with progression of the disease. PDA is intrinsically associated with biliary obstruction, infection, jaundice, ascites and pancreatic insufficiency, but beyond these factors PDA patients frequently experience the hypercatabolic state of cachexia and muscle wasting [Pausch et al. 2012]. In addition, PDA is classically associated with hypercoagulability and development of thromboembolic disease (Trousseau’s syndrome) [Khorana and Fine, 2004]. Combined with the host of unrelated ailments typical of patients in their seventh, eighth and ninth decades of life, the average condition of PDA patients is poor, and many in this population may never be eligible or receive therapy.
From an epidemiological standpoint, efforts to change the long-term outcome of PDA patients through modification of risk factors have also been disappointing. Few behaviors reliably predict an increased risk for PDA [Raimondi et al. 2009]. Of those factors, cigarette use [Iodice et al. 2008; Heinen et al. 2010] should be discouraged but others, such as dietary habits, are less definitive [Thiebaut et al. 2009] and there is not sufficient evidence to recommend dietary changes to reduce the risk of PDA. The most promising chemopreventative agent is low-dose aspirin, which has been shown to significantly reduce the risk of pancreatic cancer in a dose-dependent manner [Tan et al. 2011]. There are familial clusters of PDA [Hruban et al. 1999; Bartsch et al. 2004; Shi et al. 2009] and first-degree relatives of affected patients are at increased risk [Klein et al. 2004; Hruban et al. 2010]. However, these comprise a minority of the overall population of PDA patients (5–10%) [Bartsch et al. 2012].
Furthermore, this knowledge is of limited benefit due to the lack of validated screening tests for early diagnosis of PDA. Due to its location in the retroperitoneum, the pancreas is difficult to access and sample with traditional endoscopic techniques. Endoscopic ultrasound techniques provide for higher yields but the morbidity associated with this procedure makes it unsuitable as a screening tool in an unselected population. Studies are ongoing in targeted populations of patients at high risk [Langer et al. 2009; Verna et al. 2010; Canto et al. 2012]. Cross-sectional imaging has the potential to identify small and even asymptomatic pancreatic lesions while they are still amenable to surgical resection [Canto et al. 2012]. However, due to poor innate contrast between PDA and the surrounding pancreas, specialized imaging protocols are required to optimally image pancreatic cancer by computerized tomography (CT) and magnetic resonance imaging (MRI) [Erkan et al. 2012]. As discussed later, PDA is characterized by hypovascularity and reduced perfusion compared with normal pancreatic tissue and this property may be utilized to obtain greater resolution in the detection of early lesions using techniques such as diffusion-weighted MRI [Holzapfel et al. 2011].
Serum sampling has not yet identified a suitable screening test for early detection of PDA. Many pancreatic lesions secrete CA19-9 (carbohydrate antigen 19-9) and this serum assay has a role in some patients in monitoring disease activity and response to therapy [Steinberg, 1990]. However CA19-9 has little use alone as a screening test due to high rates of false positivity in patients with nonmalignant hepatobiliary disease [Frebourg et al. 1988]. There are ongoing efforts to identify molecular markers for early diagnosis of PDA [Goggins, 2005] but so far there are no validated agents and, as a consequence, diagnosis is often delayed.
The limited effect of local therapies
Currently, complete surgical resection provides the only potential for long-term cure of PDA but only a minority of patients have tumors that are amenable to surgery [Shaib et al. 2006]. This is due to the fact that, upon diagnosis, tumors have generally spread to involve critical abdominal vessels as well as adjacent organs. Significant advances have been made in the technical aspects of surgical resection with decreases in short-term morbidity and mortality at major centers [Winter et al. 2012]. Yet even in the most experienced centers, long-term survival after surgery is poor [Farnell et al. 2005; Ferrone et al. 2012], with tumors recurring in virtually all patients [Allison et al. 1998]. Due to the high rate of recurrence, local targeted therapy with radiation has been suggested following surgery. However, controlled studies of the long-term impact of adjuvant radiation therapy have proved inconclusive to date [Neoptolemos et al. 2004]. The cytotoxic effect of radiation therapy relies in part on the presence of oxygen [Harrison et al. 2002]. However, intraoperative oxygen measurements on human patients have found that these tumors are extremely hypoxic [Koong et al. 2000], which may contribute to the limited impact of this modality.
The limited long-term efficacy of surgery and adjuvant radiation therapy has led many to conclude that residual tumor tissue remains even in the case of complete surgical resection with no evidence of residual tumor. One possible explanation is that of ‘field effect’ mutations that may affect otherwise normal appearing cells present in the residual pancreatic tissue. Alternatively, PDA may simply metastasize at a microscopic level at a very early stage. Indeed, provocative data in a genetically engineered mouse model of PDA suggest that mutant cells may delaminate from the pancreatic epithelium and enter circulation in the very early stages of tumorigenesis even prior to the development of an overt carcinoma [Rhim et al. 2012]. If this is true, then PDA should be considered an inherently metastatic disease for which local therapy is simply a delaying action. This also highlights the importance of identifying chemotherapeutic agents that effectively target microscopic metastases. It is notable that mutation evolution analysis based on deep sequencing of human pancreatic tumor samples has suggested a long latency period for PDA, estimating that it may take an average of 17 years for a tumor to evolve from a single common progenitor [Yachida et al. 2010]. However, a computational modeling study is consistent with the notion that PDA metastasizes early in disease [Haeno et al. 2012].
Clinical data of therapeutic efforts following resection of PDA are summarized in Table 1. These trials are consistent in demonstrating a small benefit when assessing recurrence but limited impact on long-term survival regardless of the intervention [Neoptolemos et al. 2004, 2010; Stocken et al. 2005; Oettle et al. 2007]. The modest achievements of adjuvant therapy compare poorly with the experience in other common cancers. Unfortunately PDA cells display broad and intractable resistance to chemotherapy, the subject of the remainder of this review.
Table 1.
Study | Study period | Study population | Treatment arm(s) | Survival |
||
---|---|---|---|---|---|---|
Median OS (months) | p value | 5-year survival % | ||||
EORTC 40891 [Klinkenbijl et al. 1999] | 1987–1995 | Stage 1–3 resected PDA (others excluded from this analysis) | Observation | 12.6 | NS | 10 |
Chemoradiotherapy (5-FU based) | 17.1 | 20 | ||||
ESPAC-1 [Neoptolemos et al. 2004] | 1994–2000 | Stage 1–3 resected PDA | Observation | 16.9 | 0.009 (for chemo versus no chemo) | 11 |
Chemotherapy alone (5-FU) | 21.6 | 29 | ||||
Chemoradiotherapy | 13.9 | 7 | ||||
Chemoradiotherapy + chemotherapy | 19.9 | 13 | ||||
CONKO-001 [Oettle et al. 2007] | 1998–2004 | Stage 1–3, resected PDA | Observation | 20.7 | NS | 11.5 |
Chemo (gemcitabine) | 22.1 | 22.5 | ||||
RTOG 9704 [Regine et al. 2008, 2011] | 1998–2002 | Stage 1–3, resected PDA in head of pancreas. | 5-FU + chemoradiotherapy | 16.9 | NS | 22 |
Gemcitabine + chemoradiotherapy | 20.5 | 18 | ||||
ESPAC-3 [Neoptolemos et al. 2010] | 2000–2007 | Stage 1–3, resected PDA | 5-FU | 23.0 | NS | NR |
Gemcitabine | 23.6 | NR | ||||
Lung cancer | mOS | 5 year survival | ||||
ANITA [Douillard et al. 2006] | Stage IB–IIIA resected, NSCLCa | Observation | 43.7 | 0.017 | 43 | |
Chemotherapy | 65.7 | 51 | ||||
Colon cancer | 6 year survival | |||||
MOSAIC [Andre et al. 2004, 2009] | 1998–2001 | Stage II–III, resected colon cancer | Chemotherapy (FOLFOX) | NR | 78.5 | |
Breast cancer | 5 year survival | |||||
[Sparano et al. 2008] | 1999–2002 | Stage II–III breast cancer | Chemotherapy (most effective group) | NR | 89.7 |
5-FU: fluorouracil; chemo: chemotherapy; NR: not reported; NS: not significant (p > 0.05); NSCLCa: non-small cell lung cancer. OS: overall survival; PDA: pancreatic ductal adenocarcinoma.
Chemotherapy resistance in PDA
The track record of the clinical trials in advanced and metastatic pancreatic cancer is dismal (summarized in Table 2). Gemcitabine and erlotinib (Tarceva) remain the only two agents approved for use in advanced disease despite their modest benefits. Gemcitabine was approved on the basis of a study [Burris et al. 1997] showing that it was superior to 5-FU (5-fluorouracil) in providing a clinical benefit among advanced PDA patients with pain symptoms (clinical benefit rate = 23.8% versus 4.8%; p = 0.0022) and modestly prolonged median survival from 4.4 to 5.6 months (p = 0.0025). The incremental median survival benefit seen with the addition of erlotinib to gemcitabine is even smaller (5.9 to 6.2 months, p = 0.038), albeit statistically significant [Moore et al. 2007].
Table 2.
Study* | Accrual period | Number of patients | Treatment groups | Median OS |
Response rate |
||
---|---|---|---|---|---|---|---|
Months | p | % | p | ||||
[Burris et al. 1997] | 1992-1994 | 126 | 5-FU | 4.4 | 0.0025 | 0 | NS |
Gemcitabine | 5.6 | 5.4 | |||||
[Bramhall et al. 2001] | 1997-1998 | 239 | GemG + marimistat | 5.55.5 | NS | 1116 | NS |
[Moore et al. 2003] | 1997-1999 | 277 | GemG + BAY 12-9566 (MMP inhibitor) | 6.63.7 | <0.001 | NRNR | |
[Heinemann et al. 2006] | 1997-2002 | 195 | GemG + cisplatin | 6.07.5 | NS | 8.210.2 | NS |
[Berlin et al. 2002] | 1998-1999 | 322 | GemG + 5-FU | 5.4 | NS | 5.66.9 | NS |
6.7 | |||||||
[Van Cutsem et al. 2004] | 1999-2001 | 688 | Gem | 6.1 | NS | 8 | NS |
G + tipifarnib | 6.4 | 6 | |||||
[Rocha Lima et al. 2004] | 2000-2001 | 360 | Gem | 6.6 | NS | 4.1 | <0.001 |
G + irinotecan | 6.3 | 16.1 | |||||
[Louvet et al. 2005] | 2001-2003 | 313 | Gem | 7.1 | NS | 17.3 | 0.044 |
G + oxaliplatin | 9.0 | 26.8 | |||||
[Herrmann et al. 2007] | 2001-2004 | 319 | GemG + capecitabine | 7.28.4 | NS | 7.810 | NS |
[Abou-Alfa et al. 2006] | 2001-2003 | 349 | Gem | 6.2 | NS | 5.2 | NS |
G+ exatecan | 6.7 | 6.8 | |||||
[Moore et al. 2007] | 2001-2003 | 569 | Gem | 5.9 | 0.038 | 8.0 | NS |
G + erlotinib | 6.2 | 8.6 | |||||
[Oettle et al. 2005] | 2001-2003 | 565 | GemG + pemetrexed | 6.36.2 | NS | 7.114.8 | 0.004 |
[Colucci et al. 2010] | 2002-2007 | 400 | Gem | 8.3 | NS | 10.1 | NS |
G + cisplatin | 7.2 | 12.9 | |||||
[Cunningham et al. 2009] | 2002-2005 | 533 | GemG + capecitabine | 6.2 | NS | 12.4 | 0.034 |
7.1 | 19.1 | ||||||
[Poplin et al. 2009] | 2003-2005 | 832 | Gem (standard rate)Gem-FDRGem-FDR + oxaliplatin | 4.96.25.7 | NS | 6109 | NS |
[Philip et al. 2010] | 2004-2006 | 745 | GemG + cetuximab | 5.9 | NS | 7 | NS |
6.3 | 8 | ||||||
[Kindler et al. 2010] | 2004-2006 | 602 | Gem | 5.9 | NS | 10 | NS |
G + bevacizumab | 5.8 | 13 | |||||
[Van Cutsem et al. 2009] | 2005-2006 | 607 | Gem + erlotinibGem + erlotinib + bevacizumab | 6.07.1 | NS | 8.613.5 | NS |
[Conroy et al. 2011] | 2005-2009 | 342 | GemFOLFIRINOX | 6.811.1 | <0.001 | 9.431.6 | <0.001 |
[Heinemann et al. 2012] | 2006-2008 | 281 | Gem + erlotininb (capecitabine for second line)Capecitabine + erlotinib (gem for second line) | 6.26.9 | NS | 165 | NR |
[Goncalves et al. 2012] | 2006-2009 | 104 | GemG + sorafenib | 9.28 | NS | 19 | NS |
23 | |||||||
[Kindler et al. 2011] | 2007-2008 | 632 | GemG + axitinib | 8.38.5 | NS | 25 | 0.018 |
5-FU: fluorouracil; FDR: fixed dose rate.G and Gem: gemcitabine; NR: not reported; NS: not statistically significant (p > 0.05);
In addition to these trials, several phase III trials have been completed or terminated and remain unpublished. These include fluorouracil plus triacetyluridine [ClinicalTrials.gov identifier: NCT24427], aflibercept [ClinicalTrials.gov identifier: NCT574275], TS-1 [ClinicalTrials.gov identifier: NCT498225], GV1001 vaccine [ClinicalTrials.gov identifier: NCT358566], virulizin [ClinicalTrials.gov identifier: NCT40092] and AMG 479- ganitumab [ClinicalTrials.gov identifier: NCT1231347].
As summarized in Table 2, over 20 phase III trials have been conducted to improve on the modest efficacy of gemcitabine and these have been overwhelmingly disappointing. These trials covered traditional chemotherapeutic agents and combinations, targeted therapies such as the anti- vascular endothelial growth factor (anti-VEGF) monoclonal antibody, bevacizumab [Kindler et al. 2010] and the anti-epidermal growth factor receptor (anti-EGFR) antibody, cetuximab [Philip et al. 2010], as well as experimental targeted therapies including farnesyltransferase inhibitors [van Cutsem et al. 2004]. It is difficult to overstate the physical, financial and psychological costs of these unsuccessful attempts.
Two notable exceptions to this tale have been reported in the past 2 years. In 2011, a robust clinical benefit was found in a phase III randomized trial of FOLFIRINOX (a four-drug combination of 5-FU, leucovorin, oxaliplatin and irinotecan) compared with gemcitabine in metastatic PDA (median overall survival [OS] of 11.1 versus 6.8 months; p ≤ 0.001) [Conroy et al. 2011]. However, this advantage comes at the cost of significant toxicity, making the regimen appropriate only for those patients with good performance status. As recently as November 2012, after exciting phase II results [von Hoff et al. 2011], a phase III trial of nab-paclitaxel (Abraxane) plus gemcitabine was reported to have met its primary overall survival endpoint. We look forward to learning the magnitude of this effect in the coming months and are excited at the prospect of having a range of chemotherapeutic tools to treat patients in different states of health.
This collective history has delivered a consistent overarching message: the response of PDA to chemotherapy is poor. Using standard criteria that define radiographic response as a decrease of 30% (or 50% in older studies) in tumor size, very few patients treated with chemotherapy experience an objective response (noted in Table 2). Thus, the initial resistance of these tumors is primary (innate), rather than the secondary (acquired) resistance that is classically observed in most cancers. This is an important clue to understanding the recalcitrant nature of pancreatic cancer.
Cell-autonomous mechanisms of resistance to chemotherapy
It is informative to think of the resistance of PDA to chemotherapy as occurring due to cell-autonomous and non-cell-autonomous pathways. Although ductal pancreatic tumors display clinical and pathologic heterogeneity, a striking characteristic of PDA is the consistent pattern of high penetrance genetic alterations that occur in four genetic loci: K-ras, p53, cdkn2a and smad4/DPC4. Over 90% of pancreatic tumors harbor activating mutations in K-ras, one of the most potent of all human oncogenes, far exceeding the rate of any other cancer [Almoguera et al. 1988; Pellegata et al. 1994; Hezel et al. 2006; Maitra and Hruban, 2008]. Mutant K-ras initiates a signal transduction cascade that provides a strong progrowth signal, increases cell motility and invasion, and profoundly rearranges cell metabolism to a growth-promoting state. In pancreatic cancer, Ras mutation initiates paracrine signals that promote and maintain stromal desmoplasia, a key mediator of non-cell-autonomous resistance (see below). Unfortunately, K-ras is an extremely challenging therapeutic target for which no effective targeted inhibitors have been identified to date.
Overlaid on this oncogenic scaffold are mutations in four extremely potent tumor suppressor genes. The cdkn2a locus encodes two tumor suppressor genes, p16Ink4a and p15ARF. These genes are inactivated through a variety of mechanisms in >90% of human pancreatic tumors [Caldas et al. 1994; Schutte et al. 1997]. The p53 tumor suppressor gene is another major tumor suppressor and is altered in 75–90% of pancreatic tumors [Pellegata et al. 1994; Redston et al. 1994], resulting in impaired DNA damage responses, impaired apoptosis, loss of cell cycle control, and promotion of genomic instability. p53 is typically altered through ‘gain of function’ missense mutations that may further promote cancer beyond the loss of classical p53 tumor suppressor functions [Olive et al. 2004; Morton et al. 2010]. Alterations in DPC4 [Hahn et al. 1996a, 1996b] are observed in more than half of cases and confer a prometastatic phenotype. The combined effect of these mutations is formidable and likely explains a large portion of the difficulty in treating this disease. Indeed, patients with three or four of these alterations in their tumors have a much worse prognosis than those with one or two (median survival of 9 versus 23 months) [Yachida et al. 2012].
Besides these well-established ‘driver’ variations, many other genetic changes occur at lower frequencies [Hansel et al. 2003; Jones et al. 2008; Biankin et al. 2012; Perez-Mancera et al. 2012]. An effort to sequence the entire exome of 24 PDA samples revealed that the average PDA contains more than 60 genomic changes [Jones et al. 2008]. Some of these may contribute to the specific resistance to chemotherapy in as yet unidentified ways. This high degree of genomic changes seen in PDA is suggestive of significant genomic instability and may limit the effectiveness of therapy, especially targeted agents, by contributing to secondary or acquired chemoresistance.
Despite the survival benefits observed in clinical studies, only 5–10% of pancreatic tumors exhibit a radiographic response to gemcitabine therapy. Pharmacological investigations into the mechanisms of gemcitabine activity have led to some of the best characterized determinants of patient prognosis. Gemcitabine [2′,2′-difluorodeoxycytidine (dFdC)] is a nucleoside analog of cytidine that must be actively transported into cells and then sequentially phosphorylated to the active triphosphate [2′,2′-difluorodeoxycytidine triphosphate (dFdCTP)] [Heinemann et al. 1988, Mini et al. 2006]. Transport across the cell membrane is primarily mediated by human equilibrative nucleoside transporter (hENT1), though other transporters play a minor role [Mackey et al. 1998, Mini et al. 2006]. Cell lines that are resistant to gemcitabine are often hENT1 deficient [Achiwa et al. 2004] and hENT1 expression in human tissues can predict response to gemcitabine [Oguri et al. 2007]. In pancreatic cancer, patients with elevated hENT1 have improved survival when treated with gemcitabine but not among untreated patients [Marechal et al. 2012]. In a large clinical trial [Farrell et al. 2009], patients treated with gemcitabine who had no hENT1 staining had poorer survival than those with positive hENT1 staining (hazard ratio for survival = 0.51, 95% confidence interval [CI] = 0.29–0.91; p = 0.02).
Enzymes associated with the metabolic activation and inactivation of gemcitabine may also impact tumor sensitivity. The monophosphorylation of gemcitabine is a rate-limiting step in its activation and is mediated by deoxycytidine kinase (dCK). Reduced levels of dCK are associated with gemcitabine resistance in some tumor cell lines [Achiwa et al. 2004], while elevated dCK expression is associated with improved survival among those receiving adjuvant gemcitabine in PDA [Marechal et al. 2012]. Conversely, gemcitabine can be deaminated to its inactive metabolite [2′,2′-difluorodeoxyuridine (dFdU)] in a process catalyzed by the enzyme cytidine deaminase (CDA) [Eliopoulos et al. 1998], levels of which are a key determinant of gemcitabine activity. One frequent polymorphism 79A>C (Lys27Gln) is associated with decreased enzymatic activity, improved clinical outcomes, and increased toxicity in combination therapy with gemcitabine in lung cancer [Tibaldi et al. 2008]. In pancreatic cancer, the data are conflicting; one group failed to find an effect of this polymorphism on gemcitabine activity [Sugiyama et al. 2007], while another group saw increased toxicity but no change in outcomes in patients with intact CDA treated with gemcitabine [Farrell et al. 2012]. Other polymorphisms have also been identified [Sugiyama et al. 2007; Tanaka et al. 2010] and may be clinically relevant. Other studies suggest that the most relevant measure of CDA is functional testing which can predict rate of severe toxicity to gemcitabine [Ciccolini et al. 2010], though the clinical implications of these findings await validation in prospective trials [Giovannetti et al. 2010]. It is the high levels of CDA in human plasma that leads to the short (~15 minute) half-life of gemcitabine. This short half-life is compounded by non-cell-autonomous features of pancreatic tumors that limit the delivery of drugs to pancreatic tissues.
Non-cell-autonomous barriers to drug efficacy
A defining characteristic of PDA is the presence of a dense fibrotic proliferation surrounding the epithelial cells that may form the majority of the tumor mass [Chu et al. 2007; Neesse et al. 2011]. This ‘desmoplastic reaction’ is composed of various leukocytes, fibroblasts, endothelial cells and neuronal cells, as well as extracellular matrix components such as collagen and hyaluronan. The desmoplastic reaction is driven by paracrine signals originating in the epithelial compartment. These signals are driven by the oncogenic signals such as those initiated by mutant K-ras. A pair of studies in genetically engineered mouse models that utilized ‘switchable’ alleles of mutant K-ras found that the loss of mutant K-ras expression led to rapid quiescence and involution of the stroma over the course of just a few days [Collins et al. 2012; Ying et al. 2012]. In recent years, significant effort has been invested in identifying the signals that mediate the relationships between the different cell types in pancreatic tumors. For example, early in pancreatic tumor development, the neoplastic epithelial cells begin to overexpress Sonic Hedgehog (SHH), a secreted ligand that normally plays a role during organ development [Berman et al. 2003]. This upregulation has no effect on hedgehog pathway activity in the epithelial compartment [Nolan-Stevaux et al. 2009]. Rather, SHH activates the pathway in nearby stromal fibroblasts, promoting their activation and proliferation [Bailey et al. 2008; Tian et al. 2009]. This pathway has served as a paradigm for how tumor cells influence the behavior of their neighboring stromal cells.
The desmoplastic stroma of pancreatic cancer has physiological effects on the tumor that have a direct impact on drug efficacy. In contrast to many tumors that are dependent on neo-angiogenesis, ductal pancreatic tumors are very poorly vascularized relative to normal tissues and consequently poorly perfused [Olive et al. 2009]. Indirect evidence suggests that this is mediated by an anti-angiogenic effect of the tumor stroma, though the precise mechanism is an area of active research. Regardless, the poor perfusion of pancreatic tumors has the unfortunate consequence of limiting the delivery of therapeutic agents into the tumor parenchyma. Indeed, studies in a genetically engineered mouse model found that the delivery of two different chemotherapeutic agents, gemcitabine and doxorubicin, was approximately one third that of surrounding normal tissues [Olive et al. 2009]. Furthermore, poor perfusion in pancreatic tumors has been correlated with poor prognosis in patients [Komar et al. 2009]. The drug delivery effect is visualized every time a contrast agent is used to image a patient with pancreatic cancer: the finding of a ‘hypoenhancing mass’ in the pancreas is diagnostic for pancreatic ductal adenocarcinoma, particularly compared with endocrine carcinomas of the pancreas, which are hyperperfused [Fusaroli et al. 2010; Matsubara et al. 2011; Saftoiu et al. 2012]. Another consequence of poor perfusion is a hypoxic microenvironment, which can have important effects on radiosensitivity, cell metabolism and cell invasion. Direct measurements of oxygen partial pressure in human pancreatic tumors found that pancreatic tumors are profoundly hypoxic [Koong et al. 2000].
Tumor stroma is also the site of interaction between cancer and the immune system. Pancreatic tumors establish a profoundly immunosuppressed microenvironment that is nearly devoid of T lymphocytes. Several stromal cell types harbor immunosuppressive properties, including tumor associated macrophages (TAMs), cancer associated fibroblasts (CAFs), regulatory T cells (Treg) and myeloid derived suppressor cells. Recently, two groups identified a K-ras dependent signal that promotes immunosuppression. Following activation, the principle upregulated cytokine is granulocyte-macrophage colony-stimulating factor (GM-CSF), which was found to promote the recruitment and activation of immature myeloid progenitor cells to become myeloid derived suppressor cells [Bayne et al. 2012; Pylayeva-Gupta et al. 2012]. Undoubtedly, other such signals exist and should be explored in the coming years.
Future therapeutic options
The many complexities of pancreatic tumors have, to date, overwhelmed our best clinical efforts. However, the investments of the past 30 years in establishing a fundamental understanding of the disease uncovered a number of important and promising leads for new therapeutic approaches. At the most basic level, improved patient management and the advent of multidisciplinary centers specializing in the care of pancreatic cancer patients is improving the quality of life of our patients. High volume centers clearly have improved surgical outcomes [Lieberman et al. 1995; Birkmeyer et al. 2002] and similar expertise in the endoscopy suite is likely to also improve patient care. Indeed, a recent report described the diagnostic and financial advantages of direct histological processing of fine needle aspiration (FNA) samples rather than use of cytology [Brais et al. 2012]. The recent introduction of endoscopic core biopsy needles has also improved the ability to acquire samples for both diagnostic and experimental purposes.
Several promising techniques are under development that may improve diagnostic imaging. Advanced MRI sequences such as diffusion weighted imaging (DWI) and dynamic contrast enhanced (DCE) MRI capitalize on the altered perfusion of pancreatic tumor to provide functional contrast relative to normal or inflamed pancreatic tissue [Fattahi et al. 2009; Bali et al. 2011; Hur et al. 2012; Wiggermann et al. 2012; Yao et al. 2012]. Advanced endoscopic ultrasound techniques such as contrast ultrasound (which measure tissue perfusion) and ultrasound elastrography (which measures tissue stiffness) have shown initial promise as diagnostic and prognostic indicators [Sofuni et al. 2005; Sakamoto et al. 2008; D’Onofrio et al. 2009]. A number of efforts are under way to identify targeted contrast agents that discriminate early cancer or late-stage premalignancies. Among the most promising is a peptide that recognizes extracellular expression of Plectin-1, which was identified initially through a phage-display screen in genetically engineered mouse models of pancreatic cancer [Kelly et al. 2008; Bausch et al. 2009]. This probe appears to be upregulated in carcinomas in situ (PanIN 3) and is being developed for clinical trials as a single photon emission computed tomography (SPECT) probe.
For patients with advanced or metastatic PDA, a number of new therapeutic avenues are being explored. A variety of approaches have been taken to target components of the pancreatic tumor stroma. PEGPH20, a modified enzyme that breaks down hyaluronan crosslinks in the extracellular matrix, has been shown by two groups to facilitate the delivery of drugs to pancreatic tumors in genetically engineered mice and increase their overall survival [Jacobetz et al. 2013; Provenzano et al. 2012] A phase Ib/II clinical trial of PEGPH20 in combination with gemcitabine is now active at multiple sites. Two agents are in clinical trials that take advantage of the paucity of vasculature in pancreatic cancer. The gamma secretase inhibitor RO4929097 (Hoffman La Roche) is being evaluated in previously treated metastatic pancreatic cancer patients in a phase II study. Gamma-secretase is required for activation of Notch pathway signaling, which plays a role in pancreatic tumor angiogenesis. In a genetically engineered mouse model, inhibition of γ-secretase reduced vascularity to critically low levels within the tumor, resulting in cavitating necrosis and increased overall survival when combined with gemcitabine [Cook et al. 2012]. A second approach utilizes the presence of hypoxia to activate a chemotherapeutic prodrug. TH-302 is a potent DNA alkylator that is selectively activated in regions of hypoxia and is now undergoing clinical evaluation in combination with gemcitabine. Unfortunately, inhibition of the Hedgehog pathway, one of the earliest stroma targeting strategies, has so far failed to meet expectations. Two different targeted inhibitors of the Smoothened protein, IPI-926 (Saridegib, Infinity Pharmaceuticals) and GDC-0449 (Vismodegib, Genentech) were evaluated in phase II clinical trials, with negative results reported for IPI-926. Investigations are ongoing to understand this disconnect between preclinical and clinical results for these agents. On a positive note, a phase III study of gemcitabine plus Abraxane, a nanoparticle reformulation of taxol, was recently found to have met the primary endpoint, after an encouraging phase II study in which metastatic patients lived an average of 12.2 months [von Hoff et al. 2011]. One proposed mechanism of action is the targeting of SPARC, an extracellular matrix protein that is upregulated in the stroma of pancreatic tumors [Desai et al. 2009]. However, a recent analysis in genetically engineered mice found that Abraxane alters the sensitivity of pancreatic tumors to gemcitabine through downregulation of cytidine deaminase, leading to higher concentrations of dFdCTP in tumors [Frese et al. 2012]. In either case, with a toxicity profile that may be more reasonable than FOLFIRINOX, the regimen may prove to be a welcome new tool for the treatment of pancreatic cancer patients.
Finally, two novel immunotherapy approaches are under development in pancreatic cancer. The observation that GM-CSF promotes a paracrine circuit that helps maintain an immunosuppressive microenvironment has led to the proposal that anti-GM-CSF targeted antibodies may be useful in treating patients with pancreatic cancer [Bayne et al. 2012; Pylayeva-Gupta et al. 2012]. Another approach was reported in a phase I trial of a CD40 agonist in combination with gemcitabine in metastatic PDA patients. CD40 is an immunostimulant, and the combination therapy resulted in partial responses in 19% of patients and stable disease in 52% of patients. Contrary to initial expectations, the regimen relied on a macrophage-based mechanism of action, as revealed in a genetically engineered mouse model [Beatty et al. 2011].
Lessons learned
It is important to heed the hard-won lessons of a generation of clinical researchers [Tabernero and Macarulla, 2009]. As our molecular understanding of cancer in general and PDA specifically increases, it will become increasingly useful to obtain more information about the tumors we are treating. Currently many patients are diagnosed with PDA on the basis of imaging characteristics and a fine needle aspiration, which demonstrates adenocarcinoma but provides little additional tissue for further analysis. In many advanced clinical trials, the reason for failure of encouraging agents is never determined and this limits further directions in PDA research. Where therapies are developed and justified on the basis of tumor biology, it is critical to include pretreatment biopsies and, whenever feasible, to obtain additional post-treatment tumor samples to monitor the effect of targeting interventions on tumor histology and biology. Although this can increase trial costs and is an additional burden to patients, many patients believe in the importance of research efforts and are willing to undergo these procedures as a meaningful contribution to this effort. Moreover, in successful trials, correlative studies can provide valuable guidance for future development efforts. Treatment-related biopsies also facilitate the ability to prospectively test biomarkers, an important tool in identifying appropriate agents for advanced clinical trials [Philip et al. 2009].
It is also useful to bear in mind the many missed signals: situations where single arm phase I or II trials led to great hope, only to be disappointed by randomized phase III trials. In some cases this relates to the marked heterogeneity in outcomes among patients with advanced PDA based on other clinical characteristics such as age, pain status, function status and other comorbidities. These factors are complex and difficult to control. One way to address this limitation is to incorporate a control group in phase II trials to provide context to the reported results. Although this will require more patients and will still necessitate large phase III trials of positive agents [Rubinstein et al. 2011], it may reduce the number of agents that progress to expensive, large-scale, but ultimately futile phase III studies [Sharma et al. 2011].
Consideration must also be given to the caution necessary when utilizing surrogate markers in clinical trials. Particularly in pancreatic cancer, response rate and progression-free survival have often failed to correlate with increased overall survival. Although additional surrogate markers may prove to be beneficial, ultimately well-designed randomized trials with survival or clinical benefit outcomes will remain the gold standard of therapeutic effect.
In addition, it is important to remember that the majority of patients with PDA are elderly and many have poor functional status related to their tumors. These patients may ultimately benefit from different therapies than a younger, fitter population, and they should be represented in clinical trials. We therefore advocate for dedicated trials of less toxic regimens in the setting of performance status 2 patients. Perhaps most importantly, numerous advanced clinical trials have been terminated early and the data from many of these experiences are not publicly available. An effort must be made to make these data available to researchers in a timely fashion to inform future clinical trial design.
In conclusion, while the clinical outcomes for PDA have not improved sufficiently in the last decades, a large wealth of knowledge has been developed and is now being translated towards the ultimate goal of improving treatment outcomes for patients with PDA. We are filled with optimism that these efforts will be successful.
Footnotes
Funding: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
Conflict of interest statement: The authors declare no conflicts of interest in preparing this article.
Contributor Information
Paul E. Oberstein, Department of Medicine, Division of Hematology and Oncology, Columbia University Medical Center, New York, NY, USA
Kenneth P. Olive, Department of Medicine, Division of Digestive and Liver Diseases, Department of Pathology & Cell Biology, Herbert Irving Comprehensive Cancer Center, Columbia University, 1130 Saint Nicholas Ave, ICRC 217A, New York, NY 10032, USA
References
- Abou-Alfa G., Letourneau R., Harker G., Modiano M., Hurwitz H., Tchekmedyian N., et al. (2006) Randomized phase III study of exatecan and gemcitabine compared with gemcitabine alone in untreated advanced pancreatic cancer. J Clin Oncol 24: 4441–4447 [DOI] [PubMed] [Google Scholar]
- Achiwa H., Oguri T., Sato S., Maeda H., Niimi T., Ueda R. (2004) Determinants of sensitivity and resistance to gemcitabine: the roles of human equilibrative nucleoside transporter 1 and deoxycytidine kinase in non-small cell lung cancer. Cancer Sci 95: 753–757 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Allison D., Piantadosi S., Hruban R., Dooley W., Fishman E., Yeo C., et al. (1998) DNA content and other factors associated with ten-year survival after resection of pancreatic carcinoma. J Surg Oncol 67: 151–159 [DOI] [PubMed] [Google Scholar]
- Almoguera C., Shibata D., Forrester K., Martin J., Arnheim N., Perucho M. (1988) Most human carcinomas of the exocrine pancreas contain mutant C-K-Ras genes. Cell 53: 549–554 [DOI] [PubMed] [Google Scholar]
- Andre T., Boni C., Mounedji-Boudiaf L., Navarro M., Tabernero J., Hickish T., et al. (2004) Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer. N Engl J Med 350: 2343–2351 [DOI] [PubMed] [Google Scholar]
- Andre T., Boni C., Navarro M., Tabernero J., Hickish T., Topham C., et al. (2009) Improved overall survival with oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment in stage II or III colon cancer in the mosaic trial. J Clin Oncol 27: 3109–3116 [DOI] [PubMed] [Google Scholar]
- Bailey J., Swanson B., Hamada T., Eggers J., Singh P., Caffery T., et al. (2008) Sonic hedgehog promotes desmoplasia in pancreatic cancer. Clin Cancer Res 14: 5995–6004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bakkevold K., Arnesjo B., Kambestad B. (1992) Carcinoma of the pancreas and papilla of Vater: presenting symptoms, signs, and diagnosis related to stage and tumour site. A prospective multicentre trial in 472 patients. Norwegian Pancreatic Cancer Trial. Scand J Gastroenterol 27: 317–325 [DOI] [PubMed] [Google Scholar]
- Bali M., Metens T., Denolin V., Delhaye M., Demetter P., Closset J., et al. (2011) Tumoral and nontumoral pancreas: correlation between quantitative dynamic contrast-enhanced MR imaging and histopathologic parameters. Radiology 261: 456–466 [DOI] [PubMed] [Google Scholar]
- Bartsch D., Gress T., Langer P. (2012) Familial pancreatic cancer – current knowledge. Nat Rev Gastroenterol Hepatol 9: 445–453 [DOI] [PubMed] [Google Scholar]
- Bartsch D., Kress R., Sina-Frey M., Grutzmann R., Gerdes B., Pilarsky C., et al. (2004) Prevalence of familial pancreatic cancer in Germany. Int J Cancer 110: 902–906 [DOI] [PubMed] [Google Scholar]
- Bausch D., Mino-Kenudson M., Fernandez-Del Castillo C., Warshaw A., Kelly K., Thayer S. (2009) Plectin-1 is a biomarker of malignant pancreatic intraductal papillary mucinous neoplasms. J Gastrointest Surg 13: 1948–1954; discussion 1954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bayne L., Beatty G., Jhala N., Clark C., Rhim A., Stanger B., et al. (2012) Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell 21: 822–835 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beatty G., Chiorean E., Fishman M., Saboury B., Teitelbaum U., Sun W., et al. (2011) CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 331: 1612–1616 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berlin J., Catalano P., Thomas J., Kugler J., Haller D., Benson A., III. (2002) Phase III study of gemcitabine in combination with fluorouracil versus gemcitabine alone in patients with advanced pancreatic carcinoma: Eastern Cooperative Oncology Group Trial E2297. J Clin Oncol 20: 3270–3275 [DOI] [PubMed] [Google Scholar]
- Berman D., Karhadkar S., Maitra A., Montes De Oca R., Gerstenblith M., Briggs K., et al. (2003) Widespread requirement for hedgehog ligand stimulation in growth of digestive tract tumours. Nature 425: 846–851 [DOI] [PubMed] [Google Scholar]
- Biankin A., Waddell N., Kassahn K., Gingras M., Muthuswamy L., Johns A., et al. (2012) Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491: 399–405 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bilimoria K., Bentrem D., Ko C., Ritchey J., Stewart A., Winchester D., et al. (2007) Validation of the 6th edition AJCC Pancreatic Cancer Staging System: report from the National Cancer Database. Cancer 110: 738–744 [DOI] [PubMed] [Google Scholar]
- Birkmeyer J., Siewers A., Finlayson E., Stukel T., Lucas F., Batista I., et al. (2002) Hospital volume and surgical mortality in the United States. N Engl J Med 346: 1128–1137 [DOI] [PubMed] [Google Scholar]
- Brais R., Davies S., O’Donovan M., Simpson B., Cook N., Darbonne W., et al. (2012) Direct histological processing of EUS biopsies enables rapid molecular biomarker analysis for interventional pancreatic cancer trials. Pancreatology 12: 8–15 [DOI] [PubMed] [Google Scholar]
- Bramhall S., Rosemurgy A., Brown P., Bowry C., Buckels J. (2001) Marimastat as first-line therapy for patients with unresectable pancreatic cancer: a randomized trial. J Clin Oncol 19: 3447–3455 [DOI] [PubMed] [Google Scholar]
- Burris H., III, Moore M., Andersen J., Green M., Rothenberg M., Modiano M., et al. (1997) Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol 15: 2403–2413 [DOI] [PubMed] [Google Scholar]
- Caldas C., Hahn S., Da Costa L., Redston M., Schutte M., Seymour A., et al. (1994) Frequent somatic mutations and homozygous deletions of the P16 (Mts1) gene in pancreatic adenocarcinoma. Nat Genet 8: 27–32 [DOI] [PubMed] [Google Scholar]
- Canto M., Hruban R., Fishman E., Kamel I., Schulick R., Zhang Z., et al. (2012) Frequent detection of pancreatic lesions in asymptomatic high-risk individuals. Gastroenterology 142: 796–804; quiz e714–795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chari S., Leibson C., Rabe K., Ransom J., De Andrade M., Petersen G. (2005) Probability of pancreatic cancer following diabetes: a population-based study. Gastroenterology 129: 504–511 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chu G., Kimmelman A., Hezel A., Depinho R. (2007) Stromal biology of pancreatic cancer. J Cell Biochem 101: 887–907 [DOI] [PubMed] [Google Scholar]
- Ciccolini J., Dahan L., Andre N., Evrard A., Duluc M., Blesius A., et al. (2010) Cytidine deaminase residual activity in serum is a predictive marker of early severe toxicities in adults after gemcitabine-based chemotherapies.J Clin Oncol 28: 160–165 [DOI] [PubMed] [Google Scholar]
- Collins M., Bednar F., Zhang Y., Brisset J., Galban S., Galban C., et al. (2012) Oncogenic Kras is required for both the initiation and maintenance of pancreatic cancer in mice. J Clin Invest 122: 639–653 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colucci G., Labianca R., Di Costanzo F., Gebbia V., Carteni G., Massidda B., et al. (2010) Randomized phase III trial of gemcitabine plus cisplatin compared with single-agent gemcitabine as first-line treatment of patients with advanced pancreatic cancer: the Gip-1 Study. J Clin Oncol 28: 1645–1651 [DOI] [PubMed] [Google Scholar]
- Conroy T., Desseigne F., Ychou M., Bouche O., Guimbaud R., Becouarn Y., et al. (2011) Folfirinox versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 364: 1817–1825 [DOI] [PubMed] [Google Scholar]
- Cook N., Frese K., Bapiro T., Jacobetz M., Gopinathan A., Miller J., et al. (2012) Gamma secretase inhibition promotes hypoxic necrosis in mouse pancreatic ductal adenocarcinoma. J Exp Med 209: 437–444 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cunningham D., Chau I., Stocken D., Valle J., Smith D., Steward W., et al. (2009) Phase III randomized comparison of gemcitabine versus gemcitabine plus capecitabine in patients with advanced pancreatic cancer. J Clin Oncol 27: 5513–5518 [DOI] [PubMed] [Google Scholar]
- Desai N., Trieu V., Damascelli B., Soon-Shiong P. (2009) SPARC expression correlates with tumor response to albumin-bound paclitaxel in head and neck cancer patients. Transl Oncol 2: 59–64 [DOI] [PMC free article] [PubMed] [Google Scholar]
- D’Onofrio M., Zamboni G., Malago R., Mantovani W., Principe F., Gallotti A., et al. (2009) Resectable pancreatic adenocarcinoma: is the enhancement pattern at contrast-enhanced ultrasonography a pre-operative prognostic factor? Ultrasound Med Biol 35: 1929–1937 [DOI] [PubMed] [Google Scholar]
- Douillard J., Rosell R., De Lena M., Carpagnano F., Ramlau R., Gonzales-Larriba J., et al. (2006) Adjuvant vinorelbine plus cisplatin versus observation in patients with completely resected stage Ib–IIIa non-small-cell lung cancer (Adjuvant Navelbine International Trialist Association [Anita]): a randomised controlled trial. Lancet Oncol 7: 719–727 [DOI] [PubMed] [Google Scholar]
- Eliopoulos N., Cournoyer D., Momparler R. (1998) Drug resistance to 5-aza-2′-deoxycytidine, 2′,2′-difluorodeoxycytidine, and cytosine arabinoside conferred by retroviral-mediated transfer of human cytidine deaminase cDNA into murine cells. Cancer Chemother Pharmacol 42: 373–378 [DOI] [PubMed] [Google Scholar]
- Erkan M., Hausmann S., Michalski C., Fingerle A., Dobritz M., Kleeff J., et al. (2012) The Role of stroma in pancreatic cancer: diagnostic and therapeutic implications. Nat Rev Gastroenterol Hepatol 9: 454–467 [DOI] [PubMed] [Google Scholar]
- Farrell J., Bae K., Wong J., Guha C., Dicker A., Elsaleh H. (2012) Cytidine deaminase single-nucleotide polymorphism is predictive of toxicity from gemcitabine in patients with pancreatic cancer: RTOG 9704. Pharmacogenomics J 12: 395–403 [DOI] [PubMed] [Google Scholar]
- Farnell M., Pearson R., Sarr M., Dimagno E., Burgart L., Dahl T., et al. (2005) A Prospective randomized trial comparing standard pancreatoduodenectomy with pancreatoduodenectomy with extended lymphadenectomy in resectable pancreatic head adenocarcinoma. Surgery 138: 618–628; discussion 628–630. [DOI] [PubMed] [Google Scholar]
- Farrell J., Elsaleh H., Garcia M., Lai R., Ammar A., Regine W., et al. (2009) Human equilibrative nucleoside transporter 1 levels predict response to gemcitabine in patients with pancreatic cancer. Gastroenterology 136: 187–195 [DOI] [PubMed] [Google Scholar]
- Fattahi R., Balci N., Perman W., Hsueh E., Alkaade S., Havlioglu N., et al. (2009) Pancreatic diffusion-weighted imaging (DWI): comparison between mass-forming focal pancreatitis (FP), pancreatic cancer (PC), and normal pancreas. J Magn Reson Imaging 29: 350–356 [DOI] [PubMed] [Google Scholar]
- Ferrone C., Pieretti-Vanmarcke R., Bloom J., Zheng H., Szymonifka J., Wargo J., et al. (2012) Pancreatic ductal adenocarcinoma: long-term survival does not equal cure. Surgery 152(Suppl.): S43–S49 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frebourg T., Bercoff E., Manchon N., Senant J., Basuyau J., Breton P., et al. (1988) The evaluation of Ca 19-9 antigen level in the early detection of pancreatic cancer. A prospective study of 866 patients. Cancer 62: 2287–2290 [DOI] [PubMed] [Google Scholar]
- Frese K., Neesse A., Cook N., Bapiro T., Lolkema M., Jodrell D., et al. (2012) nab-Paclitaxel potentiates gemcitabine activity by reducing cytidine deaminase levels in a mouse model of pancreatic cancer. Cancer Discov 2: 260–269 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fusaroli P., Spada A., Mancino M., Caletti G. (2010) Contrast harmonic echo-endoscopic ultrasound improves accuracy in diagnosis of solid pancreatic masses. Clin Gastroenterol Hepatol 8: 629–634, e621–622 [DOI] [PubMed] [Google Scholar]
- Giovannetti E., Tibaldi C., Falcone A., Danesi R., Peters G. (2010) Impact of cytidine deaminase polymorphisms on toxicity after gemcitabine: the question is still ongoing. J Clin Oncol 28: e221–222; author reply e223–225. [DOI] [PubMed] [Google Scholar]
- Goggins M. (2005) Molecular markers of early pancreatic cancer. J Clin Oncol 23: 4524–4531 [DOI] [PubMed] [Google Scholar]
- Goncalves A., Gilabert M., Francois E., Dahan L., Perrier H., Lamy R., et al. (2012) Baypan Study: a double-blind phase III randomized trial comparing gemcitabine plus sorafenib and gemcitabine plus placebo in patients with advanced pancreatic cancer. Ann Oncol 23: 2799–2805 [DOI] [PubMed] [Google Scholar]
- Haeno H., Gonen M., Davis M., Herman J., Iacobuzio-Donahue C., Michor F. (2012) Computational modeling of pancreatic cancer reveals kinetics of metastasis suggesting optimum treatment strategies. Cell 148: 362–375 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hahn S., Hoque A., Moskaluk C., Da Costa L., Schutte M., Rozenblum E., et al. (1996a) Homozygous deletion map at 18q21.1 in pancreatic cancer. Cancer Res 56: 490–494 [PubMed] [Google Scholar]
- Hahn S., Schutte M., Hoque A., Moskaluk C., Da Costa L., Rozenblum E., et al. (1996b) DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science 271: 350–353 [DOI] [PubMed] [Google Scholar]
- Hansel D., Kern S., Hruban R. (2003) Molecular Pathogenesis of pancreatic cancer. Annu Rev Genomics Hum Genet 4: 237–256 [DOI] [PubMed] [Google Scholar]
- Harrison L., Chadha M., Hill R., Hu K., Shasha D. (2002) Impact of tumor hypoxia and anemia on radiation therapy outcomes. Oncologist 7: 492–508 [DOI] [PubMed] [Google Scholar]
- Heinemann V., Hertel L., Grindey G., Plunkett W. (1988) Comparison of the cellular pharmacokinetics and toxicity of 2′,2′-difluorodeoxycytidine and 1-beta-D-arabinofuranosylcytosine. Cancer Res 48: 4024–4031 [PubMed] [Google Scholar]
- Heinemann V., Quietzsch D., Gieseler F., Gonnermann M., Schonekas H., Rost A., et al. (2006) Randomized phase III trial of gemcitabine plus cisplatin compared with gemcitabine alone in advanced pancreatic cancer. J Clin Oncol 24: 3946–3952 [DOI] [PubMed] [Google Scholar]
- Heinemann V., Vehling-Kaiser U., Waldschmidt D., Kettner E., Marten A., Winkelmann C., et al. (2012) Gemcitabine plus erlotinib followed by capecitabine versus capecitabine plus erlotinib followed by gemcitabine in advanced pancreatic cancer: final results of a randomised phase III trial of the ‘Arbeitsgemeinschaft Internistische Onkologie’ (AIO-PK0104). Gut, in press. DOI: 10.1136/gutjnl-2012-302759 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heinen M., Verhage B., Goldbohm R., Van Den Brandt P. (2010) Active and passive smoking and the risk of pancreatic cancer in the Netherlands Cohort Study. Cancer Epidemiol Biomarkers Prev 19: 1612–1622 [DOI] [PubMed] [Google Scholar]
- Herrmann R., Bodoky G., Ruhstaller T., Glimelius B., Bajetta E., Schuller J., et al. (2007) Gemcitabine plus capecitabine compared with gemcitabine alone in advanced pancreatic cancer: a randomized, multicenter, phase III trial of the Swiss Group for Clinical Cancer Research and the Central European Cooperative Oncology Group. J Clin Oncol 25: 2212–2217 [DOI] [PubMed] [Google Scholar]
- Hezel A., Kimmelman A., Stanger B., Bardeesy N., Depinho R. (2006) Genetics and Biology of pancreatic ductal adenocarcinoma. Genes Dev 20: 1218–1249 [DOI] [PubMed] [Google Scholar]
- Holzapfel K., Reiser-Erkan C., Fingerle A., Erkan M., Eiber M., Rummeny E., et al. (2011) Comparison of diffusion-weighted MR imaging and multidetector-row CT in the detection of liver metastases in patients operated for pancreatic cancer. Abdom Imaging 36: 179–184 [DOI] [PubMed] [Google Scholar]
- Hruban R., Canto M., Goggins M., Schulick R., Klein A. (2010) Update on familial pancreatic cancer. Adv Surg 44: 293–311 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hruban R., Petersen G., Goggins M., Tersmette A., Offerhaus G., Falatko F., et al. (1999) Familial pancreatic cancer. Ann Oncol 10(Suppl. 4): 69–73 [PubMed] [Google Scholar]
- Hur B., Lee J., Lee J., Park J., Kim S., Joo I., et al. (2012) Magnetic resonance imaging findings of the mass-forming type of autoimmune pancreatitis: comparison with pancreatic adenocarcinoma. J Magn Reson Imaging 36: 188–197 [DOI] [PubMed] [Google Scholar]
- Iodice S., Gandini S., Maisonneuve P., Lowenfels A. (2008) Tobacco and the risk of pancreatic cancer: a review and meta-analysis. Langenbecks Arch Surg 393: 535–545 [DOI] [PubMed] [Google Scholar]
- Jacobetz M., Chan D., Neesse A., Bapiro T., Cook N., Frese K., et al. (2013) Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut 62: 112–120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jemal A., Bray F., Center M., Ferlay J., Ward E., Forman D. (2011) Global cancer statistics. CA Cancer J Clin 61: 69–90 [DOI] [PubMed] [Google Scholar]
- Jones S., Zhang X., Parsons D., Lin J., Leary R., Angenendt P., et al. (2008) Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 321: 1801–1806 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelly K., Bardeesy N., Anbazhagan R., Gurumurthy S., Berger J., Alencar H., et al. (2008) Targeted nanoparticles for imaging incipient pancreatic ductal adenocarcinoma. PLoS Med 5: e85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelsen D., Portenoy R., Thaler H., Niedzwiecki D., Passik S., Tao Y., et al. (1995) Pain and depression in patients with newly diagnosed pancreas cancer. J Clin Oncol 13: 748–755 [DOI] [PubMed] [Google Scholar]
- Khorana A., Fine R. (2004) Pancreatic cancer and thromboembolic disease. Lancet Oncol 5: 655–663 [DOI] [PubMed] [Google Scholar]
- Kindler H., Ioka T., Richel D., Bennouna J., Letourneau R., Okusaka T., et al. (2011) Axitinib plus gemcitabine versus placebo plus gemcitabine in patients with advanced pancreatic adenocarcinoma: a double-blind randomised phase III study. Lancet Oncol 12: 256–262 [DOI] [PubMed] [Google Scholar]
- Kindler H., Niedzwiecki D., Hollis D., Sutherland S., Schrag D., Hurwitz H., et al. (2010) Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: phase III trial of the Cancer and Leukemia Group B (CALGB 80303). J Clin Oncol 28: 3617–3622 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klein A., Brune K., Petersen G., Goggins M., Tersmette A., Offerhaus G., et al. (2004) Prospective risk of pancreatic cancer in familial pancreatic cancer kindreds. Cancer Res 64: 2634–2638 [DOI] [PubMed] [Google Scholar]
- Klinkenbijl J., Jeekel J., Sahmoud T., van Pel R., Couvreur M., Veenhof C., et al. (1999) Adjuvant radiotherapy and 5-fluorouracil after curative resection of cancer of the pancreas and periampullary region: phase III TRIAL of the EORTC Gastrointestinal Tract Cancer Cooperative Group. Ann Surg 230: 776–782; discussion 782–774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Komar G., Kauhanen S., Liukko K., Seppanen M., Kajander S., Ovaska J., et al. (2009) Decreased blood flow with increased metabolic activity: a novel sign of pancreatic tumor aggressiveness. Clin Cancer Res 15: 5511–5517 [DOI] [PubMed] [Google Scholar]
- Koong A., Mehta V., Le Q., Fisher G., Terris D., Brown J., et al. (2000) Pancreatic tumors show high levels of hypoxia. Int J Radiat Oncol Biol Phys 48: 919–922 [DOI] [PubMed] [Google Scholar]
- Langer P., Kann P., Fendrich V., Habbe N., Schneider M., Sina M., et al. (2009) Five years of prospective screening of high-risk individuals from families with familial pancreatic cancer. Gut 58: 1410–1418 [DOI] [PubMed] [Google Scholar]
- Lieberman M., Kilburn H., Lindsey M., Brennan M. (1995) Relation of perioperative deaths to hospital volume among patients undergoing pancreatic resection for malignancy. Ann Surg 222: 638–645 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Louvet C., Labianca R., Hammel P., Lledo G., Zampino M., Andre T., et al. (2005) Gemcitabine in combination with oxaliplatin compared with gemcitabine alone in locally advanced or metastatic pancreatic cancer: results of a gercor and giscad phase III trial. J Clin Oncol 23: 3509–3516 [DOI] [PubMed] [Google Scholar]
- Mackey J., Mani R., Selner M., Mowles D., Young J., Belt J., et al. (1998) Functional nucleoside transporters are required for gemcitabine influx and manifestation of toxicity in cancer cell lines. Cancer Res 58: 4349–4357 [PubMed] [Google Scholar]
- Maitra A., Hruban R. (2008) Pancreatic cancer. Annu Rev Pathol 3: 157–188 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marechal R., Bachet J., Mackey J., Dalban C., Demetter P., Graham K., et al. (2012) Levels of gemcitabine transport and metabolism proteins predict survival times of patients treated with gemcitabine for pancreatic adenocarcinoma. Gastroenterology 143: 664–674 e661–666 [DOI] [PubMed] [Google Scholar]
- Matsubara H., Itoh A., Kawashima H., Kasugai T., Ohno E., Ishikawa T., et al. (2011) Dynamic quantitative evaluation of contrast-enhanced endoscopic ultrasonography in the diagnosis of pancreatic diseases. Pancreas 40: 1073–1079 [DOI] [PubMed] [Google Scholar]
- Mini E., Nobili S., Caciagli B., Landini I., Mazzei T. (2006) Cellular pharmacology of gemcitabine. Ann Oncol 17(Suppl. 5): v7–12 [DOI] [PubMed] [Google Scholar]
- Moore M., Goldstein D., Hamm J., Figer A., Hecht J., Gallinger S., et al. (2007) Erlotinib Plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 25: 1960–1966 [DOI] [PubMed] [Google Scholar]
- Moore M., Hamm J., Dancey J., Eisenberg P., Dagenais M., Fields A., et al. (2003) Comparison of gemcitabine versus the matrix metalloproteinase inhibitor BAY 12-9566 in patients with advanced or metastatic adenocarcinoma of the pancreas: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 21: 3296–3302 [DOI] [PubMed] [Google Scholar]
- Morton J., Timpson P., Karim S., Ridgway R., Athineos D., Doyle B., et al. (2010) Mutant P53 drives metastasis and overcomes growth arrest/senescence in pancreatic cancer. Proc Natl Acad Sci U S A 107: 246–251 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neesse A., Michl P., Frese K., Feig C., Cook N., Jacobetz M., et al. (2011) Stromal biology and therapy in pancreatic cancer. Gut 60: 861–868 [DOI] [PubMed] [Google Scholar]
- Neoptolemos J., Stocken D., Bassi C., Ghaneh P., Cunningham D., Goldstein D., et al. (2010) Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: a randomized controlled trial. JAMA 304: 1073–1081 [DOI] [PubMed] [Google Scholar]
- Neoptolemos J., Stocken D., Friess H., Bassi C., Dunn J., Hickey H., et al. (2004) A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med 350: 1200–1210 [DOI] [PubMed] [Google Scholar]
- Nolan-Stevaux O., Lau J., Truitt M., Chu G., Hebrok M., Fernandez-Zapico M., et al. (2009) Gli1 is regulated through smoothened-independent mechanisms in neoplastic pancreatic ducts and mediates pdac cell survival and transformation. Genes Dev 23: 24–36 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oettle H., Post S., Neuhaus P., Gellert K., Langrehr J., Ridwelski K., et al. (2007) Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled Trial. JAMA 297: 267–277 [DOI] [PubMed] [Google Scholar]
- Oettle H., Richards D., Ramanathan R., van Laethem J., Peeters M., Fuchs M., et al. (2005) A phase III trial of pemetrexed plus gemcitabine versus gemcitabine in patients with unresectable or metastatic pancreatic cancer. Ann Oncol 16: 1639–1645 [DOI] [PubMed] [Google Scholar]
- Oguri T., Achiwa H., Muramatsu H., Ozasa H., Sato S., Shimizu S., et al. (2007) The absence of human equilibrative nucleoside transporter 1 expression predicts nonresponse to gemcitabine-containing chemotherapy in non-small cell lung cancer. Cancer Lett 256: 112–119 [DOI] [PubMed] [Google Scholar]
- Olive K., Jacobetz M., Davidson C., Gopinathan A., Mcintyre D., Honess D., et al. (2009) Inhibition of hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324: 1457–1461 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olive K., Tuveson D., Ruhe Z., Yin B., Willis N., Bronson R.T., et al. (2004) Mutant p53 gain of function in two mouse models of Li-Fraumeni syndrome. Cell 119: 847–860 [DOI] [PubMed] [Google Scholar]
- Pausch T., Hartwig W., Hinz U., Swolana T., Bundy B., Hackert T., et al. (2012) Cachexia but not obesity worsens the postoperative outcome after pancreatoduodenectomy in pancreatic cancer. Surgery 152: S81–88 [DOI] [PubMed] [Google Scholar]
- Pellegata N., Sessa F., Renault B., Bonato M., Leone B., Solcia E., et al. (1994) K-ras and p53 gene mutations in pancreatic cancer: ductal and nonductal tumors progress through different genetic lesions. Cancer Res 54: 1556–1560 [PubMed] [Google Scholar]
- Perez-Mancera P., Rust A., van der Weyden L., Kristiansen G., Li A., Sarver A., et al. (2012) The deubiquitinase USP9X suppresses pancreatic ductal adenocarcinoma. Nature 486: 266–270 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Philip P., Benedetti J., Corless C., Wong R., O’Reilly E., Flynn P., et al. (2010) Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group-Directed Intergroup Trial S0205. J Clin Oncol 28: 3605–3610 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Philip P., Mooney M., Jaffe D., Eckhardt G., Moore M., Meropol N., et al. (2009) Consensus report of the national cancer institute clinical trials planning meeting on pancreas cancer treatment. J Clin Oncol 27: 5660–5669 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poplin E., Feng Y., Berlin J., Rothenberg M., Hochster H., Mitchell E., et al. (2009) Phase III, randomized study of gemcitabine and oxaliplatin versus gemcitabine (fixed-dose rate infusion) compared with gemcitabine (30-minute infusion) in patients with pancreatic carcinoma E6201: a trial of the Eastern Cooperative Oncology Group. J Clin Oncol 27: 3778–3785 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Porta M., Fabregat X., Malats N., Guarner L., Carrato A., De Miguel A., et al. (2005) Exocrine pancreatic cancer: symptoms at presentation and their relation to tumour site and stage. Clin Transl Oncol 7: 189–197 [DOI] [PubMed] [Google Scholar]
- Provenzano P., Cuevas C., Chang A., Goel V., von Hoff D., Hingorani S.R. (2012) Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 21: 418–429 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pylayeva-Gupta Y., Lee K., Hajdu C., Miller G., Bar-Sagi D. (2012) Oncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasia. Cancer Cell 21: 836–847 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raimondi S., Maisonneuve P., Lowenfels A.B. (2009) Epidemiology of pancreatic cancer: an overview. Nat Rev Gastroenterol Hepatol 6: 699–708 [DOI] [PubMed] [Google Scholar]
- Redston M., Caldas C., Seymour A., Hruban R., Da Costa L., Yeo C., et al. (1994) P53 mutations in pancreatic carcinoma and evidence of common involvement of homocopolymer tracts in DNA microdeletions. Cancer Res 54: 3025–3033 [PubMed] [Google Scholar]
- Regine W., Winter K., Abrams R., Safran H., Hoffman J., Konski A., et al. (2008) Fluorouracil vs gemcitabine chemotherapy before and after fluorouracil-based chemoradiation following resection of pancreatic adenocarcinoma: a randomized controlled trial. JAMA 299: 1019–1026 [DOI] [PubMed] [Google Scholar]
- Regine W., Winter K., Abrams R., Safran H., Hoffman J., Konski A., et al. (2011) Fluorouracil-based chemoradiation with either gemcitabine or fluorouracil chemotherapy after resection of pancreatic adenocarcinoma: 5-year analysis of the U.S. Intergroup/RTOG 9704 phase III trial. Ann Surg Oncol 18: 1319–1326 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rhim A., Mirek E., Aiello N., Maitra A., Bailey J., Mcallister F., et al. (2012) EMT and dissemination precede pancreatic tumor formation. Cell 148: 349–361 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rocha Lima C., Green M., Rotche R., Miller W., Jr, Jeffrey G., Cisar L., et al. (2004) Irinotecan plus gemcitabine results in no survival advantage compared with gemcitabine monotherapy in patients with locally advanced or metastatic pancreatic cancer despite increased tumor response rate. J Clin Oncol 22: 3776–3783 [DOI] [PubMed] [Google Scholar]
- Rubinstein L., Leblanc M., Smith M. (2011) More randomization in phase II trials: necessary but not sufficient. J Natl Cancer Inst 103: 1075–1077 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saftoiu A., Dietrich C., Vilmann P. (2012) Contrast-enhanced harmonicendoscopic ultrasound. Endoscopy 44: 612–617 [DOI] [PubMed] [Google Scholar]
- Sakamoto H., Kitano M., Suetomi Y., Maekawa K., Takeyama Y., Kudo M. (2008) Utility of contrast-enhanced endoscopic ultrasonography for diagnosis of small pancreatic carcinomas. Ultrasound Med Biol 34: 525–532 [DOI] [PubMed] [Google Scholar]
- Schutte M., Hruban R., Geradts J., Maynard R., Hilgers W., Rabindran S., et al. (1997) Abrogation of the Rb/p16 tumor-suppressive pathway in virtually all pancreatic carcinomas. Cancer Res 57: 3126–3130 [PubMed] [Google Scholar]
- Shaib Y., Davila J., El-Serag H. (2006) The epidemiology of pancreatic cancer in the United States: changes below the surface. Aliment Pharmacol Ther 24: 87–94 [DOI] [PubMed] [Google Scholar]
- Sharma M., Stadler W., Ratain M. (2011) Randomized phase II trials: a long-term investment with promising returns. J Natl Cancer Inst 103: 1093–1100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi C., Hruban R., Klein A. (2009) Familial pancreatic cancer. Arch Pathol Lab Med 133: 365–374 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siegel R., Naishadham D., Jemal A. (2012) Cancer statistics, 2012. CA Cancer J Clin 62: 10–29 [DOI] [PubMed] [Google Scholar]
- Sofuni A., Iijima H., Moriyasu F., Nakayama D., Shimizu M., Nakamura K., et al. (2005) Differential diagnosis of pancreatic tumors using ultrasound contrast imaging. J Gastroenterol 40: 518–525 [DOI] [PubMed] [Google Scholar]
- Sparano J., Wang M., Martino S., Jones V., Perez E., Saphner T., et al. (2008) Weekly paclitaxel in the adjuvant treatment of breast cancer. N Engl J Med 358: 1663–1671 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stathis A., Moore M. (2010) Advanced pancreatic carcinoma: current treatment and future challenges. Nat Rev Clin Oncol 7: 163–172 [DOI] [PubMed] [Google Scholar]
- Steinberg W. (1990) The clinical utility of the CA 19-9 tumor-associated antigen. Am J Gastroenterol 85: 350–355 [PubMed] [Google Scholar]
- Stocken D., Buchler M., Dervenis C., Bassi C., Jeekel H., Klinkenbijl J., et al. (2005) Meta-analysis of randomised adjuvant therapy trials for pancreatic cancer. Br J Cancer 92: 1372–1381 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sugiyama E., Kaniwa N., Kim S., Kikura-Hanajiri R., Hasegawa R., Maekawa K., et al. (2007) Pharmacokinetics of gemcitabine in japanese cancer patients: the impact of a cytidine deaminase polymorphism. J Clin Oncol 25: 32–42 [DOI] [PubMed] [Google Scholar]
- Tabernero J., Macarulla T. (2009) Changing the paradigm in conducting randomized clinical studies in advanced pancreatic cancer: an opportunity for better clinical development. J Clin Oncol 27: 5487–5491 [DOI] [PubMed] [Google Scholar]
- Tan X., Reid Lombardo K., Bamlet W., Oberg A., Robinson D., Anderson K., et al. (2011) Aspirin, nonsteroidal anti-inflammatory drugs, acetaminophen, and pancreatic cancer risk: a clinic-based case-control study. Cancer Prev Res 4: 1835–1841 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanaka M., Javle M., Dong X., Eng C., Abbruzzese J., Li D. (2010) Gemcitabine metabolic and transporter gene polymorphisms are associated with drug toxicity and efficacy in patients with locally advanced pancreatic cancer. Cancer 116: 5325–5335 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thiebaut A., Jiao L., Silverman D., Cross A., Thompson F., Subar A., et al. (2009) Dietary fatty acids and pancreatic cancer in the NIH-AARP Diet and Health Study. J Natl Cancer Inst 101: 1001–1011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tian H., Callahan C., Dupree K., Darbonne W., Ahn C., Scales S., et al. (2009) Hedgehog signaling is restricted to the stromal compartment during pancreatic carcinogenesis. Proc Natl Acad Sci U S A 106: 4254–4259 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tibaldi C., Giovannetti E., Vasile E., Mey V., Laan A., Nannizzi S., et al. (2008) Correlation of CDA, ERCC1, and XPD polymorphisms with response and survival in gemcitabine/cisplatin-treated advanced non-small cell lung cancer patients. Clin Cancer Res 14: 1797–1803 [DOI] [PubMed] [Google Scholar]
- Van Cutsem E., van de Velde H., Karasek P., Oettle H., Vervenne W., Szawlowski A., et al. (2004) Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J Clin Oncol 22: 1430–1438 [DOI] [PubMed] [Google Scholar]
- Van Cutsem E., Vervenne W., Bennouna J., Humblet Y., Gill S., van Laethem J., et al. (2009) Phase III trial of bevacizumab in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer. J Clin Oncol 27: 2231–2237 [DOI] [PubMed] [Google Scholar]
- Verna E., Hwang C., Stevens P., Rotterdam H., Stavropoulos S., Sy C., et al. (2010) Pancreatic cancer screening in a prospective cohort of high-risk patients: a comprehensive strategy of imaging and genetics. Clin Cancer Res 16: 5028–5037 [DOI] [PubMed] [Google Scholar]
- Von Hoff D., Ramanathan R., Borad M., Laheru D., Smith L., Wood T., et al. (2011) Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. J Clin Oncol 29: 4548–4554 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wiggermann P., Grutzmann R., Weissenbock A., Kamusella P., Dittert D., Stroszczynski C. (2012) Apparent diffusion coefficient measurements of the pancreas, pancreas carcinoma, and mass-forming focal pancreatitis. Acta Radiol 53: 135–139 [DOI] [PubMed] [Google Scholar]
- Winter J., Brennan M., Tang L., D’Angelica M., Dematteo R., Fong Y., et al. (2012) Survival after resection of pancreatic adenocarcinoma: results from a single institution over three decades. Ann Surg Oncol 19: 169–175 [DOI] [PubMed] [Google Scholar]
- Yachida S., Jones S., Bozic I., Antal T., Leary R., Fu B., et al. (2010) Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 467: 1114–1117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yachida S., White C., Naito Y., Zhong Y., Brosnan J., Macgregor-Das A., et al. (2012) Clinical significance of the genetic landscape of pancreatic cancer and implications for identification of potential long term survivors. Clin Cancer Res 18: 6339–6347 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yao X., Zeng M., Wang H., Sun F., Rao S., Ji Y. (2012) Evaluation of pancreatic cancer by multiple breath-hold dynamic contrast-enhanced magnetic resonance imaging at 3.0T. Eur J Radiol 81: e917–922 [DOI] [PubMed] [Google Scholar]
- Ying H., Kimmelman A., Lyssiotis C., Hua S., Chu G., Fletcher-Sananikone E., et al. (2012) Oncogenic Kras maintains pancreatic tumors through regulation of anabolic glucose metabolism. Cell 149: 656–670 [DOI] [PMC free article] [PubMed] [Google Scholar]